14
Hindawi Publishing Corporation e Scientific World Journal Volume 2013, Article ID 732340, 13 pages http://dx.doi.org/10.1155/2013/732340 Review Article Dendrimeric Systems and Their Applications in Ocular Drug Delivery Burçin Yavuz, Sibel BozdaL Pehlivan, and NurGen Ünlü Pharmaceutical Technology Department, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey Correspondence should be addressed to Sibel Bozda˘ g Pehlivan; [email protected] Received 7 August 2013; Accepted 9 September 2013 Academic Editors: A. Concheiro and J. M. Reid Copyright © 2013 Burc ¸in Yavuz et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Ophthalmic drug delivery is one of the most attractive and challenging research area for pharmaceutical scientists and ophthalmologists. Absorption of an ophthalmic drug in conventional dosage forms is seriously limited by physiological conditions. e use of nonionic or ionic biodegradable polymers in aqueous solutions and colloidal dosage forms such as liposomes, nanoparticles, nanocapsules, microspheres, microcapsules, microemulsions, and dendrimers has been studied to overcome the problems mentioned above. Dendrimers are a new class of polymeric materials. e unique nanostructured architecture of dendrimers has been studied to examine their role in delivery of therapeutics and imaging agents. Dendrimers can enhance drug’s water solubility, bioavailability, and biocompatibility and can be applied for different routes of drug administration successfully. Permeability enhancer properties of dendrimers were also reported. e use of dendrimers can also reduce toxicity versus activity and following an appropriate application route they allow the delivery of the drug to the targeted site and provide desired pharmacokinetic parameters. erefore, dendrimeric drug delivery systems are of interest in ocular drug delivery. In this review, the limitations related to eye’s unique structure, the advantages of dendrimers, and the potential applications of dendrimeric systems to ophthalmology including imaging, drug, peptide, and gene delivery will be discussed. 1. Introduction Drug delivery to the eye is still one of the most challenging tasks for pharmaceutical scientists. e eye is characterized with its complex structure with high resistance to drugs as well as other foreign substances. e anterior and posterior segments of the eye function both independently upon an ocular application [1]. us, ocular drug delivery can be classified into anterior and posterior segments. Conventional drug delivery systems are not effective enough to meet the requirements in the treatment of ocular diseases [2]. However, “more than 90% of the marketed ophthalmic formulations” are in the form of eye drops, and most of them target the “anterior segment eye diseases” [3]. Poor bioavailability of drugs from ocular dosage forms is mainly due to the “precorneal loss factors” including solution drainage, lacrimation, tear dynamics, tear dilution, tear turnover, conjunctival absorption, transient residence time in the cul-de-sac, and low permeability of the corneal epithelial membrane which are the major challenges to ante- rior segment drug delivery following topical administration. Treatment of “posterior segment diseases” still remains as an unsolved issue. Most of the ophthalmic diseases affect neu- ral retina, choroid, and vitreous. For example, glaucoma, dia- betic retinopathy, age-related macular degeneration (AMD), and various forms of retinitis pigmentosa are damaging the posterior eye segment, which may cause impaired vision and even blindness [4]. Delivery of drugs to the posterior segment is more challenging than to the anterior segment, due to the acellular nature of the vitreous body and the longer diffusion distance [5]. us, posterior eye segment diseases have become an important therapeutic target with unmet medical needs. e major goal in the treatment of posterior segments diseases is the delivery of therapeutic doses of drugs to the tissues while reducing the effects. Systems developed to achieve this goal range from simple solutions to novel drug delivery systems, such as nanoparticles, liposomes, micelles, dendrimers, iontophoresis, and gene delivery systems [57].

Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

Hindawi Publishing CorporationThe Scientific World JournalVolume 2013, Article ID 732340, 13 pageshttp://dx.doi.org/10.1155/2013/732340

Review ArticleDendrimeric Systems and Their Applications inOcular Drug Delivery

Burçin Yavuz, Sibel BozdaL Pehlivan, and NurGen Ünlü

Pharmaceutical Technology Department, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey

Correspondence should be addressed to Sibel Bozdag Pehlivan; [email protected]

Received 7 August 2013; Accepted 9 September 2013

Academic Editors: A. Concheiro and J. M. Reid

Copyright © 2013 Burcin Yavuz et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Ophthalmic drug delivery is one of the most attractive and challenging research area for pharmaceutical scientists andophthalmologists. Absorption of an ophthalmic drug in conventional dosage forms is seriously limited by physiological conditions.The use of nonionic or ionic biodegradable polymers in aqueous solutions and colloidal dosage forms such as liposomes,nanoparticles, nanocapsules, microspheres, microcapsules, microemulsions, and dendrimers has been studied to overcome theproblems mentioned above. Dendrimers are a new class of polymeric materials. The unique nanostructured architecture ofdendrimers has been studied to examine their role in delivery of therapeutics and imaging agents. Dendrimers can enhance drug’swater solubility, bioavailability, and biocompatibility and can be applied for different routes of drug administration successfully.Permeability enhancer properties of dendrimers were also reported. The use of dendrimers can also reduce toxicity versus activityand following an appropriate application route they allow the delivery of the drug to the targeted site and provide desiredpharmacokinetic parameters.Therefore, dendrimeric drug delivery systems are of interest in ocular drug delivery. In this review, thelimitations related to eye’s unique structure, the advantages of dendrimers, and the potential applications of dendrimeric systemsto ophthalmology including imaging, drug, peptide, and gene delivery will be discussed.

1. Introduction

Drug delivery to the eye is still one of the most challengingtasks for pharmaceutical scientists. The eye is characterizedwith its complex structure with high resistance to drugs aswell as other foreign substances. The anterior and posteriorsegments of the eye function both independently upon anocular application [1]. Thus, ocular drug delivery can beclassified into anterior and posterior segments.

Conventional drug delivery systems are not effectiveenough to meet the requirements in the treatment of oculardiseases [2]. However, “more than 90% of the marketedophthalmic formulations” are in the form of eye drops, andmost of them target the “anterior segment eye diseases”[3]. Poor bioavailability of drugs from ocular dosage formsis mainly due to the “precorneal loss factors” includingsolution drainage, lacrimation, tear dynamics, tear dilution,tear turnover, conjunctival absorption, transient residencetime in the cul-de-sac, and low permeability of the corneal

epithelial membrane which are the major challenges to ante-rior segment drug delivery following topical administration.

Treatment of “posterior segment diseases” still remains asan unsolved issue.Most of the ophthalmic diseases affect neu-ral retina, choroid, and vitreous. For example, glaucoma, dia-betic retinopathy, age-related macular degeneration (AMD),and various forms of retinitis pigmentosa are damaging theposterior eye segment, which may cause impaired visionand even blindness [4]. Delivery of drugs to the posteriorsegment is more challenging than to the anterior segment,due to the acellular nature of the vitreous body and the longerdiffusion distance [5]. Thus, posterior eye segment diseaseshave become an important therapeutic target with unmetmedical needs. The major goal in the treatment of posteriorsegments diseases is the delivery of therapeutic doses of drugsto the tissues while reducing the effects. Systems developed toachieve this goal range from simple solutions to novel drugdelivery systems, such as nanoparticles, liposomes, micelles,dendrimers, iontophoresis, and gene delivery systems [5–7].

Page 2: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

2 The Scientific World Journal

Dendrimers are “tree-like,” nanostructured polymers thathave been interesting in terms of ocular drug delivery. Theyare attractive systems for drug delivery due to their nanosizerange, ability to display multiple surface groups that allowsfor targeting, and easy preparation and functionalization [8].Ongoing studies in developing improved ocular dendrimericsystems may not only serve to enhance the drug delivery tothe ocular surface, but also may provide effective delivery oftherapeutic agents to intraocular tissues, such as the retina orchoroid, using noninvasive delivery methods.

2. Challenges in Ocular Drug Delivery

Eye has a unique physical structure with protective barriers,which offers many challenges to the effective delivery ofdrugs to the eye. The eyeball is divided into 2 segments:the anterior segment containing the cornea, crystalline lens,iris, ciliary body, and fluid-filled aqueous humor and theposterior segment that includes the sclera, choroid vessels,retina, macula, optic nerve, and fluid-filled vitreous humor[2]. This organ is well protected with various specializedcellular modifications that give rise to various barriers thatpartially isolate the eye from the rest of the body, whichcan be a challenge for drug delivery [9]. These specialprocesses/barriers are as follows.

(i) The “inner and outer blood–retinal barriers” sepa-rate the retina and the vitreous from the systemiccirculation, and because of the absence of the cellularcomponents in vitreous body, it reduces convection ofmolecules [10].

(ii) The inner limiting membrane controls the exchangeand entry of particles from the vitreous to the retina.

(iii) The “blood-aqueous barrier” limits the transport ofmolecules from the blood to the inner part of the eye[11].

(iv) Intact structure of corneal epithelium with desmo-somes and tight junctions offers resistance to thepassage of most drugs due to the presence of lay-ers: hydrophobic epithelium, hydrophilic stroma, andhydrophobic endothelium [12].

(v) The tear film forms a mucoaqueous barrier thatcontinuously washes away the particles at the anteriorsurface of the eye [9].

The anatomical and physiological barriers mentionedabove are a challenge to ocular drug delivery. Solubility,lipophilicity, molecular size and shape, charge, and degreeof ionization of the drug also affect the penetration rateto the eye [13]. Drug delivery systems’ biocompatibility isalso relevant when ocular delivery is concerned. The specificchallenge of designing an ocular therapeutic system is toachieve an efficient concentration of the drug at the activesite for the duration to provide the therapeutic efficacy [14,15]. The requirements of an ideal topical formulation tothe eye must be fulfilled as follows: the formulation mustbe well tolerated and easy to administer; avoid systemicabsorption and increase drug retention time in the eye.

Various ophthalmic formulations have been developed toimprove ocular penetration, reduce toxicity, and improvetolerability [16, 17].

Typically “less than 5% of the topically applied drug”penetrates the cornea and reaches intraocular tissues, whilemost of the instilled dose is often absorbed systemically viathe nasolacrimal duct and conjunctiva [3]. The eye dropsare easy to instill but only a very small amount of theinstilled dose is absorbed into the target tissues. It becomesnecessary to apply large doses of drugs frequently to achievethe effective therapeutic dose which leads to an increase inboth local and systemic side effects [18]. Since the penetra-tion to cornea is often poor, either systemic or intravitrealadministration (injection or implant) is required in order totreat posterior segment diseases. Due to strong blood-oculartissue barrier, systemic administration requires large doses,while intravitreal injections and implants are very invasiveand are associated with a high degree of retinal damage risk,such as retinal detachment and endophthalmitis [19]. Thus,there has been growing attention to transscleral route in orderto deliver drug to the back of the eye [20]. Sclera is morepermeable than cornea and even it is highly permeable tothe large molecules of even protein size. However, it is morecomplicated to deliver the drug to retina, because in case oftransscleral application the drugmust pass across the choroidand retina pigment epithelium (RPE) [21].

The major goal is to develop suitable drug deliverysystems with improved bioavailability of drugs, increasedretention time, reduced side effects, cellular targeting, bet-ter patient compliance, and providing extended therapeuticeffects [22]. Currently, nanocarrier-based ocular drug deliv-ery systems including dendrimers appear to be the mostpromising way to meet the requirements of an ideal oculardrug delivery system.

3. Dendrimer Structure, Synthesis,and Properties

Dendrimers are monodisperse macromolecules with severalreactive end groups that surround a small molecule andform an internal cavity. Their tree-like branched architec-ture displaying a variety of controlled terminal groups isin particular very promising for biomedical applications[23]. Especially low generation dendrimers can encapsulatehydrophobic drug molecules into their internal cavities.Because of this unique structure, dendrimers are able tosolubilize poorly water-soluble drugs [24]. In addition to theextraordinary structural control, another outstanding featureof dendrimers is their actual mimicry of globular proteins.They are referred to as “artificial proteins,” based on theirsystematic, electrophoretic, dimensional length scaling andother biomimetic properties [25, 26].

Elements are added to dendrimer structure by a chemicalreaction series and build a branching spherical structure froma starting atom such as nitrogen. The central core moleculeshould have at least two reactive functional groups and therepeated branches are organized in a series of “radicallyconcentric layers” called “generations” [27]. A schematic

Page 3: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 3

representation of a generation 2 dendrimer is given inFigure 1. Dendrimers are generally prepared using either adivergent method or a convergent one [28]. In the divergentmethod, dendrimer grows outwards from a multifunctionalcoremolecule. On the other hand, in the convergent approach,the dendrimer is constructed stepwise, starting from theend groups and progressing inwards. When the branchedpolymeric arms (dendrons) grew enough, they are attachedto a multifunctional core molecule [29]. A schematic repre-sentation of divergent and convergent methods, is given inFigure 2.Other approaches have been developed based on thedivergent and convergentmethods such as double exponentialgrowth, lego chemistry, and click chemistry. Preparation ofmonomers from a single starting material for both divergentand convergentmethods is possible using double exponentialgrowth approach.Then two result products are reacted to givea trimer, which can be used to repeat the growth again [30]. Inlego chemistry strategy, phosphorus dendrimers are preparedfrom highly functionalized cores and branched monomers.After several variations in general synthetic scheme, a schemeis developed that allows multiplications of the number ofterminal surface groups from “48 to 250” in one step [31].

Compared to other polymers, dendrimers have so manyadvantages such as their nanosize ranging from 1 to 100 nmwith lower polydispersity index that allows them to avoidRES uptake. Furthermore, targeting anywhere in the bodyis also possible, thanks to the multiple functional groupson their surface which makes it possible to attach vectordevices [32, 33]. Dendrimers have the ability to encapsu-late drug molecules into their internal cavities which leadsto enhanceed solubility, permeability, and retention effectdepending on their molecular weight. It was reported thatdrug absorption is increased with dendrimers association inthe cationic> uncharged> anionic order, where cationic den-drimers show permeation enhancement due to their abilityof interacting lipid bilayers. Smaller generation dendrimersalso have an enhancer effect on permeability since they havea better ability to move between cells [34].

Dendrimer cytotoxicity is related to the core chem-istry; the nature of the dendrimer surface is the mostinfluencing factor, because the interaction between surfacecationic charge of dendrimers and negatively charged bio-logical membranes is the main reason of toxicity. Lowergeneration dendrimers with anionic or neutral polar surfacegroups were reported to have higher biocompatibility ascompared to higher generation dendrimers with neutralapolar and cationic surface groups. It was reported thatfollowing repeated intravenous use or topical ocular appli-cation, dendrimers with cationic end groups are often toxic,whereas anionic dendrimers are not.Thus, in order to reducetoxicity, it is necessary to modify the surface amine groups ofthese dendrimers with neutral or anionic moieties [35–37].Recently, several studies have been published to report thatocular dendrimeric formulations were developed withoutcytotoxicity or irritation [38, 39]. For ocular drug delivery,it is very important to make sure that the dendrimers aresafe because serious side effects may occur due to cyto-toxicity at the ocular tissues. Safe dendrimer formulationsfor ocular drug delivery should have properties such as

Molecular cargo space

Core

Surface group

Branches

Figure 1: Schematic representation of a generation 2 dendrimer.

(A)

(B)

(a)(b)

2x (a) 3x (b)

16x8x4x

Figure 2: Schematic representation of divergent and convergentmethods: (A) the divergent growth method (B) the convergentgrowth method.

biocompatibility and low immunogenicity; thus they shouldbe carefully designed and evaluated. Furthermore, in order toovercome the potential toxicity of the dendrimers, it is veryimportant for ophthalmologists to participate and contributeto the scientific process alongside with chemists, formulationscientists and engineers.

4. Types of Dendrimers

The first “dendrimer family” that was synthesized, character-ized, and commercialized was poly(amidoamine) (PAMAM)dendrimers (Figure 3(a)) which were synthesized by the“divergent” method [25]. The structure of PAMAM den-drimers starts from an ammonia (NH

3) or ethylenediamine

(C2H8N2) molecule as a core that binds to the amine groups

of branches (R-NH2) and amide (–CONH

2R). Dendrimers

growth reaches a critical point where the branching armslimit their development into higher generations due to stericeffect that starts with G7. This effect decreases the syntheticyields of generations between G7 and G10 and prohibitsthe synthesis of any larger dendrimers [40, 41]. PAMAMdendrimers have a size range between 1.1 and 12.4 nm as theirgenerations grow through 1–10 [42]. These dimensions havebeen compared to proteins (3–8 nm), linear polymer-drugconjugates (5–20 nm), and viruses (25–240 nm). Overall,

Page 4: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

4 The Scientific World Journal

H2N

H2N

H2N

H2N

H2N

H2N

H2N

H2N

H2N

NH

NH

NH

NH

NH

NHNH

NH

NH

NH NH

NH

NH

NH

NHNH

NH

NH

NH

NH

NH

O

OO

O

O

O

O

O

O

O

O

O

OO

OOO

OO

O

O

N

N

N

N

N

N

N

N

N

N

NH2

NH2

NH2

(a)

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2H2NCH2CH2

CH2CH2CH2CH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH2

CH2CH2CH2NH

N

NN

N

NN

NN

N

N

N

N

N

N

2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2

CH2CH2CH2CH2CH2CH2

(b)

O

O

O

O

OO

O

OHHO

HO

OH

OH

CH3(OCH2CH2)22O

CH3(OCH2CH2)22O

CH3(OCH2CH2)22O

O(CH2CH2)22CH3

O(CH2CH2)22CH3

(c)

NH

O

NH

HN

O

NH

HN

HNO

O O

O

O

Core

NHNH2

NH2

NH2

NH2

NH2

NH2

NH2

NH2

(d)

Figure 3: Structures of various types of dendrimers: (a) PAMAM dendrimer, (b) polypropyleneimine dendrimer, (c) polyaryl etherdendrimer, and (d) biodegradable polylysine dendron.

PAMAMdendrimers are considered as ideal carriers for drugdelivery due to their high aqueous solubility, large varietyof surface groups, and unique architecture. For medicalapplications, the most widely studied PAMAM dendrimershave been the derivatives with an −NH

2surface, a −COOH

surface, and an –OH surface [43].Poly(amidoamine) organosilicon (PAMAMOS) dendri-

mers are silicon containing first commercial dendrimerswhich are inverted unimolecular micelles that contain exte-rior hydrophobic organosilicon and interiorly hydrophilic,nucleophilic polyamidoamine [44].

Polypropyleneimine (PPI) dendrimers (Figure 3(b)) havebeen investigated for their medical applications, but it hasbeen shown that the presence of multiple cationic aminegroups causes significant toxicity. These dendrimers aregenerally poly-alkyl amines with primary amine end groupsand they are commercially available up to generation 5[45]. Polyaryl ether dendrimers (Figure 3(c)) also have beenevaluated for drug delivery, but it was found that it is requiredto use solubilizing groups at the periphery of them due totheir poor water solubility [46].

In addition, biodegradable dendrimers have beendesigned such as those based on polylysine (Figure 3(d)),poly(disulfide amine), polyether, or polyester and after

suitable surface modifications they have been developed aspromising antiviral, antibacterial, chemotherapeutic, andvaccine carrier candidates. Glycodendrimers, that includecarbohydrates in their architecture, also have great potentialas drug carriers.Most of the glycodendrimers have saccharideresidues on their outer surface, but glycodendrimerswith a sugar central core have also been described [47].Amino acid-based dendrimers, peptide dendrimers,hydrophobic dendrimers, and asymmetric dendrimers werealso investigated for a variety of pharmaceutical applications[30, 48].

Surface engineered dendrimers were developed as a strat-egy for abatement of dendrimer toxicity. Functionalizationhelps the dendrimers gain some beneficial properties fortheir use as a drug delivery system as well as reducing theinherent toxicity [37]. One of themost popular modificationsof dendrimer surface is PEGylation which offers so manyadvantages in addition to cytotoxicity reduction such asimproved bioavailability/oral delivery application related toimproved biodistribution and pharmacokinetics, enhancedsolubility, increase in drug loading, sustained and controlleddelivery of drugs, better transfection efficiency, and tumorlocalization [49]. Acetylation is another surface engineeringapproach to reduce toxicity based on modification of surfaceamino groups with acetyl groups [50].

Page 5: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 5

(a) (b)

Figure 4: Schematic representation of drug encapsulated (a) and drug conjugated dendrimers (b).

There are already several dendrimer-based FDA-approved products in the market. For example, StratusCS Acute Care (Dade Behring), containing dendrimer-linked monoclonal antibody, was launched for “cardiacdiagnostic testing,” while another product based onmodified“Tomalia-type PAMAM” dendrimers, SuperFect (Qiagen),is a well-known gene transfection agent available for a widerange of cell lines [51, 52]. In addition, VivaGel, which is aformulation of “polyanionic lysine G4 dendrimers” with ananionic surface of “naphthalenedisulfonate (SPL7013) in aCarbopol gel” that shows antiviral activity against HIV andHSV for the treatment, has already been taken into clinicaltrials by Starpharma, according to FDA requirements. It iscurrently in Phase III clinical trials and it is also the subjectof a license agreement with Durex condoms for use as acondom coating [53, 54].

5. Interactions between Dendrimers andDrug Molecules

The dendrimer end group functionality can be modifiedto obtain molecules with novel biological properties suchas cooperative receptor-ligand interactions, which will helpdendrimers to interact with poorly soluble drugs. Den-drimers are able to increase the cellular uptake, bioavail-ability, and therapeutic efficacy, and they can also be usedto optimize the biodistribution and to reduce the systemictoxicity, clearance, and degradation rate drugs [55].

There are twomethods of dendrimer drug delivery: eitherlipophilic drugs can be encapsulated within the hydrophobicdendrimer cavity to make themwater soluble or drugs can becovalently attached onto the dendrimer surface. Encapsula-tion traps the drug inside the dendrimer using the interactionbetween drug and the dendrimer or the steric bulk of theexterior of the dendrimer. The nature of drug encapsulationmay be either a simple physical entrapment or can involvenonbonding-specific interactions within the dendrimer. Onthe other hand, the drug is attached to the exterior of

the dendrimer in dendrimer/drug conjugates. Conjugates areusually prodrugs that are either inactive or have decreasedactivity. The covalent conjugation of the drugs was mainlyused for targeting and achieving the higher drug payload,whereas the noncovalent interactions have resulted in highersolubility of insoluble drugs [23, 56]. A basic schematicrepresentation of drug encapsulated and drug conjugateddendrimers is given in Figure 4. The unique architecturalfeature of dendrimers and dendrons makes them also idealfor the construction of cross-linked covalent gels, and for theself-assembled noncovalent gels [57].

Drug dendrimer interactions are affected by the structure,generation, concentration, and surface engineering of thedendrimers. For example, PAMAM and PPI have a slightlydifferent dendritic framework. This difference in the internalbranchesmakes PPI dendrimers relativelymore hydrophobiccompared to PAMAM dendrimers and that results in dif-ferent solubilizing power [58]. Furthermore, modification ofdendrimer surface can improve the therapeutic efficacy ofdrugs in terms of targeting and reduced toxicity.

5.1. Encapsulation of Drugs within Dendritic Structure. Theacid-base reaction between the dendrimers and the guestmolecules such as drugs with coulomb attractions pulls theguest molecules inside the dendrimer structure, whereas thehydrogen bonding keeps them together.

Jansen and coworkers reported the first encapsulationof a dye inside a dendrimer in 1994, the so-called “dendritic box” [59]. Guest molecules could be entrapped inthe dendritic cavities during the synthetic process, withthe help of a shell preventing diffusion from the struc-tures, even after prolonged heating, sonication, or solventextraction [60, 61]. Following encapsulation of dyes todendrimers, anticancer drug encapsulation was the focusof the research. Kojima et al. encapsulated the anticancerdrugs methotrexate and doxorubicin using G3 and G4ethylenediamine-based poly(amidoamine) (PAMAM) den-drimers with poly(ethyleneglycol) monomethyl ether (M-PEG) grafts [62]. The same group also attached methotrexate

Page 6: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

6 The Scientific World Journal

and folic acid to the exterior of the dendritic structure andtargeted the tumor cells using drug-dendrimer conjugates[63].

Dendrimers with an apolar core and polar shell have beenreferred to as “unimolecular micelles,” whereas the dendriticstructure is independent of dendrimer concentration unlikeconventional micelles [64]. However, this approach has adisadvantage that it is difficult to control the release ofdrugs from the dendrimer core. Poly(ethylene glycol) (PEG)has been used to modify dendrimers by conjugating PEGto the dendrimer surface to form a unimolecular micelleby providing a hydrophilic shell around the dendritic core.PEGylated dendrimers are of particular interest in drugdelivery because they are biocompatible and highly watersoluble and they have ability to modify the biodistribution ofcarrier systems [65, 66].

Zimmerman et al. synthesized “cored dendrimers” withmodified dendritic architecture to encapsulate the drug.Following the synthesis, the core was removed via cleavageof ester bonds, while the rest of the structure remained thesame as a consequence of robust ether linkages [67, 68].

“Dendrimeric block copolymers” have been synthesizedwith linear hydrophilic blocks and a hydrophobic dendriticblock and their ability to complex “poorly water solu-ble” molecules have been investigated. A series of “G1–G5PAMAM-block-PEG-block-PAMAM triblock copolymers”were synthesized by Kim et al. and studied as potentialpolymeric gene carrier [69].

Dendrimer-mediated complexation has advantages interms of stability, release control, high drug loading, andlower toxicity of entrapped drugs. However, the noncovalentcomplexation often leads to lower drug encapsulation andcomplex stability compared to covalent conjugation [70].

5.2. Dendrimer Drug Conjugation. The outer surfaces ofdendrimers have been investigated as potential interactionsites with drugs to increase the loading capacity. The numberof available surface groups for drug interactions increases intwofolds with each higher generation of dendrimer. Drugscan be conjugated to dendritic systems through ester, amide,or some other linkage depending on dendritic surface, whichcan be hydrolysed by endosomal or lysosomal enzymes,inside the cell [55, 56]. There are several ionizable groupson the surface of dendrimers, where ionizable drugs canattach electrostatically.Themain used points of attachment toconjugate drugs to dendrimers are amides, esters, disulfides,hydrazones, thiol-maleimide, and sulfinyl [43]. Many reportson drug loaded-dendrimers have shown that the release ofthe free drug can be enhanced by a suitable linker choice,especially, the linker/spacer length and flexibility. Some ofthe linkers are pH-sensitive and have proven to enhanceintracellular release of the free drug [71].

Patri et al. compared the covalently conjugated drug andnoncovalent inclusion complex in terms of release kineticsand efficacy, using generation 5 PAMAM dendrimers for tar-geting methotrexate. This study indicated that the inclusioncomplex releases the drug immediately and drug is active invitro, while covalently conjugated drug is better suited for

specifically targeted drug delivery [72]. A greater control overdrug release can be achieved by the covalent drug attach-ment using biodegradable linkages than electrostatic drugs-dendrimer complexes. However, the major disadvantage ofthe conjugation is the possibility of less active drug release ortoo slow drug liberation potential to be effective in vivo [73].

5.3. Dendritic Gels. Pharmaceutical applications of hydro-gels have attracted attention because of their architecturalproperties, drug loading capacity, and controlled drug releasecapability. Hydrogels are hydrophilic and “three-dimensionalpolymeric networks” have found application in drug deliverydue to their high water absorbing capacity [74]. “In situ form-ing gels” have been investigated for a variety of applicationsincluding ocular, oral, nasal, vaginal, rectal, and injectable.[75, 76]. Dendrimers and dendrons’ can be prepared withcontrolled molecular sizes due to their dendritic structureand their nature is between traditional gel polymers and theorganic compounds with low molecular weight used in the“self assembled supramolecular gels” [57].

A “polymer network” is usually obtained with the use ofa crosslinker during polymerization. Synthesis of hydrogelshas been a function of themultivalent crosslinker behavior ofdendriticmolecules [77]. It has been shown that the dendriticbranching has an important role in the self-assembly control.Furthermore, the repeated use of its key structural motifscan lead to multiple interactions between branched units andstrengthen the noncovalent interactions responsible for the“self-assembly process” [78].

6. Ocular Applications of Dendrimeric DrugDelivery Systems

Dendrimers have been investigated for ophthalmic drugdelivery since it offers a number of advantages as a carriersystem. It has been reported that dendrimers were usedfor several purposes such as drug delivery, gene delivery,antioxidant delivery, peptide delivery, biomedical imaging,and genetic testing in ophthalmology [79]. A list of the ocularapplications of dendrimers is given in Table 1.

Dendrimers are able to transport into and out of the cells.PAMAM dendrimers can have different cell entry pathways,depending on the functional groups on the surface. AnionicPAMAM dendrimers are endocytosed primarily througha caveolin-mediated process, whereas neutral and cationicdendrimers are internalizated in cells following a clathrin-mediated process. These pathways can be highly beneficial inthe case of crossing the epithelial and retinal barriers in thecornea and retina [80, 81].

Different ocular application routes have been success-fully used for dendrimeric drug delivery and better watersolubility, permeability, bioavailability, and biocompatibilityhave been reported. Vandamme and Brobeck have evaluatedseveral series of poly(amidoamine) (PAMAM) dendrimersfor controlled ocular drug delivery and residence time ofpilocarpine nitrate and tropicamide was found to be longerfor the anionic dendrimer solutions. Results of a “mioticactivity test” on albino rabbits showed that these PAMAM

Page 7: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 7Ta

ble1:Oculara

pplications

ofdend

rimersa

nddend

rimericdeliverysyste

ms.

Drug

Dendrim

ertype

Administratio

nTreatm

ent

Outcomes

References

Piocarpine

nitratea

ndtro

picamide

PAMAM

G1.5

-4To

pical

Myosis

andmydria

sisIncreasedcornealresidence

and

prolon

gedredu

ctionof

IOP

[82]

Carteolol

Phosph

orus

containing

dend

rimers

Topical

Glau

coma

Increasedcornealresidence

and

redu

cedtoxicityandIO

P[83]

Gatifloxacin

Dendrim

ericpo

lygu

anidilyated

translo

cators

Topical

Con

junctiv

itisa

ndintraocular

infections

Enhanced

cornealtranspo

rtand

increasedantim

icrobialactiv

ity[84]

Glucosaminea

ndglucosam

ine6

-sulfate

PAMAM

G3.5-CO

OH

Subcon

junctiv

alinjection

Antiang

iogenicinglaucoma

surgery

Redu

cedinflammationandno

scar

form

ation

[38]

Carbop

latin

PAMAM

G3.5-CO

OH(dendrim

eric

nano

particles

)Subcon

junctiv

alinjection

Retin

oblasto

ma

Increasedhalflifea

ndbioavailability

Redu

ceddrug

toxicityandtumor

mass

[39]

VEG

F-ODN

Lipo

philica

mino-acid

dend

rimer

Intravitrealinjectio

nCN

VProlon

gedsupp

ressionof

VEG

Fandneovasculariz

ation

[85]

—Po

lyprop

yleneimineo

ctaamineG

2Cornealscaffold

Cornealtissuee

ngineerin

gEn

hanced

human

cornealepithelial

cellgrow

th[86]

—Surfa

cemod

ified-C

OOHending

dend

rimers

Cornealscaffold

Cornealtissuee

ngineerin

gProm

oted

adhesio

nand

proliferatio

nof

human

corneal

epith

elialcells

[87]

—Mod

ified

G1,G2,andG3dend

rimers

Topical(cornealhydrogel

adhesiv

e)Cornealwou

nds

Wou

ndsealingandno

scar

form

ation

[88]

Photosensitizer

G3aryletherd

endrim

erzinc

porphyrin

Intravenou

sinjection-ph

otod

ynam

ictheraphy

CNV

Accumulationin

neovasculariz

edarea

[89]

Con

canavalin

APo

rphyrin

glycod

endrim

ers

Topical-P

hotodynamictherapy

Intraoculartum

orsa

ndretin

oblasto

ma

Enhanced

targetingandredu

ced

toxicity

[90]

—Po

rphyrin

dend

rimers

Topical-P

hotodynamictherapy

AMDandCN

VSelectivea

ccum

ulationin

inflammatorycells

andprolon

ged

retentiontim

e[91,92]

DNA

Phthalocyanine

dend

rimers

Accumulationin

photo-irr

adiated

areasa

ndincreasedtransgene

expressio

n[93]

—Anion

icandcatio

nicc

arbo

silane

dend

rimers

Topical

Tolerance

Hydrogenbo

ndingbetweenmucin

andPA

MAM-enh

ancedretention

time

[94]

Puerarin

PAMAM

Topical

Ocularh

ypertensionand

cataract

Increasedbioavailability

[95]

Fluo

cino

lone

aceton

ide

PAMAM

G4-OH

Intravitrealinjectio

nRe

tinalneuroinfl

ammation

Redu

cedinflammation

[96]

Brim

onidinea

ndtim

olol

maleate

PAMAM

hydrogel(G

3)To

pical

Glaucom

aIncreasedup

take

[97]

Brim

onidinea

ndtim

olol

maleate

Hybrid

PAMAM

dend

rimer

hydrogel/

PLGAnano

particle

Topical

Glaucom

aIncreasedup

take

[98]

—Lys xCy

s ydend

riticpo

lymers-in

situgel

Topical

Cataractincisio

nsWou

ndsealing

[99]

Page 8: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

8 The Scientific World Journal

formulations improved pilocarpine nitrate bioavailabilitycompared to the control and also prolonged the reduction ofintraocular pressure (IOP), indicating increased precornealresidence time [82].

Durairaj et al. investigated dendrimeric polyguanidily-ated translocators (DPTs), which are a class of dendrimerswith tritolyl branches and surface guanidine groups as poten-tial ophthalmic carriers for gatifloxacin, a “fourth generationfluoroquinolone” approved for conjunctivitis treatment. Theresults have indicated that the DPT forms stable gatifloxacincomplexes and enhances solubility, permeability, anti-MRSAactivity, and in vivo delivery of gatifloxacin and seems like apotential delivery system allowing once a day dosing [84].

“Phosphorus containing dendrimers,” with one quater-nary ammonium salt as core and several carboxylic acidterminal groups have been synthesized from generation 0to generation 2 by Spataro et al. These dendrimers havebeen tested in vivo in a rabbit model to evaluate ocularcarteolol delivery and an increase of the carteolol amount inthe aqueous humour is observed. No irritation was observed,even several hours after cationic dendrimers were applied[83].

Shaunak et al. have synthesized water soluble conjugatesof D(+)-glucosamine and D(+)-glucosamine 6-sulfate withanionic PAMAM (G3.5) dendrimers to obtain synergistic“immunomodulatory and antiangiogenic effect.” When den-drimer glucosamine and dendrimer glucosamine 6-sulfateconjugates were used together in a clinically relevant scar tis-sue formation rabbit model after glaucoma filtration surgery,it has been shown that the long-term success of the surgeryhas increased from 30% to 80%. Furthermore, neither micro-bial infections nor clinical, biochemical, or hematologicaltoxicity was observed in all animals [38].

Intraocular tumors such as retinoblastoma present a highrisk of complications with high metastatic potential. One ofthe limited studies that have been done for drug deliveryto intraocular tumors has explored the use of PAMAMdendrimers with carboxyl end groups (G3.5-COOH) forextended half life and sustained delivery of carboplatin withlowered therapeutic toxicity. Carboplatin-loaded PAMAMdendrimer complexes were explored in a transgenic murineretinoblastoma model, following subconjunctival adminis-tration. It was reported that the carboplatin-loaded den-drimer nanoparticles not only crossed the sclera, but werealso retained for an extended period of time in the tumorvasculature, providing a sustained treatment effect [39].

In another research, biocompatible conjugates oflipophilic amino-acid dendrimers have been developed withcollagen scaffolds to obtain better physical and mechanicalproperties and adhesion ability. Dendrimers-based approachwas used for antivascular endothelial growth factoroligonucleotide (VEGF-ODN) delivery and successfullytested in a rat model to treat choroidal neovascularization(CNV). The results indicated that dendrimer/ODN-1complexes significantly suppressed VEGF expression in celllevel studies around 40 to 60%. Examinations of injectedrat eyes also showed that no significant toxicity and damagewere caused by the complex injections [85].

The repair of wounds such as corneal wounds that arisefrom surgical procedures has a significant importance forclinical aspects and research. Therefore, Duan and cowork-ers have generated highly crosslinked collagen using G2polypropyleneimine octaamine dendrimers to use it as atissue-engineering corneal scaffold. The optical transparen-cies of the dendrimer crosslinked collagen, EDC (1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide hydrochloride),and glutaraldehyde crosslinked collagen thermal gels werecompared and the transparency of dendrimer crosslinkedcollagen was found to be significantly higher. Results haveshown that dendrimer crosslinked collagen gels improved“human corneal epithelial cell growth” and adhesion withoutcell toxicity [86]. The same group conjugated the “surfacemodified dendrimers” with cell adhesion peptides to beused as corneal tissue engineering scaffolds and the materialhas been incorporated into both bulk structures of the gelsand onto the gel surface. Dendrimer amine groups weremodified using carboxyl group and it was found that thesurface modification promoted human corneal epithelial celladhesion and proliferation [87].

Grinstaff has developed a series of dendrimeric adhesives,to repair corneal wounds, composed of generations 1, 2,and 3 (G1, G2, and G3) combined with PEG, glycerol, andsuccinic acid. The polymer was modified to contain ter-minal methacrylate groups, ([G1]-PGLSA-MA)

2-PEG. Two

strategies have been explored to form the ocular adhesives:a photocrosslinking reaction and a peptide ligation reactionto couple the individual dendrimers together. It was reportedthat both hydrogels were adhesive, elastic, soft, transparent,and hydrophilic. The in vivo studies in chicken eyes haveindicated that the photocrosslinkable ([G1]-PGLSA-MA)

2-

PEG adhesive completely sealed on postoperative day. Thehistological studies have also demonstrated that woundssealed using these adhesive gels appeared to be more com-plete after 28 days as compared to sutured wounds. Theadvantage of photo-cross-linked gels is the light-inducedability of the polymer to crosslink and adhere to the tissue;however, there is a potential risk of ocular damagewhenusinglight [88].

Photodynamic therapy is a potentially efficient treat-ment for retinoblastoma along with the other varioussolid tumours. Makky et al. have designed a photosensi-tizer, porphyrin-based glycodendrimers with the mannose-specific ligand protein Concanavalin A conjugated on totheir surface, to specifically target the tumor cells in theretina. It was reported that the mannosylated dendrimersdemonstrated specific interactions with the receptors in thelipid bilayer and accumulation in malignant ocular tissuewas enhanced [90]. Dendrimers have also been exploredas drug carriers and photosensitizers for exudative age-related macular degeneration (AMD) and CNV treatment.Nishiyama et al. investigated porphyrin-based dendrimersfor their efficacy in treating retinal tumors and exudativeAMD associated with CNV. The formulations showed selec-tive accumulation within 24 h in the CNV lesions wheninjected into a CNV rat model [91, 92]. The same groupdeveloped phthalocyanine core-based dendrimer photosen-sitizers, which can be used to compact and deliver therapeutic

Page 9: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 9

genes with a targeting approach. Transgene expression wasmonitored only in the irradiated areas upon subconjunctivalinjection of the dendrimer formulation and followed by laserirradiation [93].

Sugisaki et al. investigated the accumulation of den-drimer porphyrin (DP), DP encapsulated polymericmicelles,and the efficacy of photodynamic therapy (PDT) using acorneal neovascularization model in mice. In this study a3rd generation “aryl ether dendrimer zinc porphyrin” withcarboxyl ending groups and polymeric micelles composedof the DP and PEG-poly(L-lysine) was used for PDT as aphotosensitizer formulation. The results showed that follow-ing administration, in 1 hour to 24 hours, both DP and DP-micelle were accumulated in the neovascularized area [89].

Bravo-Osuna and coworkers investigated the in vitroand in vivo tolerance of carbosilane dendrimers (G1 andG3, anionic and cationic) for topical ophthalmic adminis-tration. Formulations were applied to New Zealand albinorabbits and it was reported that animals did not presenteither discomfort or clinical signs after the administrationof dendrimer solutions. Nonionic interactions via hydrogenbonding between the PAMAM dendrimer surface moietiesandmucins were observed.MTT test results also showed thatanionic dendrimers were nontoxic for both conjunctival andcorneal cells [94].

In a recent study, Puerarin–dendrimer complexes wereprepared using PAMAM dendrimers (G3.5, G4, G4.5, andG5) and their physicochemical properties, in vitro release,corneal permeation, and ocular residence times were deter-mined. Valia-Chien evaluated the corneal permeation andocular residence time in rabbits using diffusion cells withexcised corneas. It was reported that puerarin-dendrimercomplexes exhibited longer residence time in rabbit eyes thanpuerarin eye drops, without damage to the corneal epitheliumor endothelium. Also results of the in vitro release studiesshowed that puerarin release was much more slower fromcomplexes than the free puerarin in PBS. However, cornealpermeation studies suggested that there was no significantdifference between puerarin-dendrimer complexes and puer-arin eye drops on drug permeability coefficient [95].

Targeted drug therapy for retinal neuroinflammationwas explored using “G4.0 hydroxyl-terminated PAMAMdendrimer-drug conjugate nanodevices” by Iezzi et al. Fluo-cinolone acetonide was conjugated to the dendrimers and invivo efficacy was assessed for over a 4-week period, using the“Royal College of Surgeons rat retinal degenerationmodel.” Itwas reported that upon intravitreal administration PAMAMdendrimers were selectively localized within “activated outerretinal microglia” in two retinal degeneration rat models andthe dendrimers were detected in the target cells, even 35 daysafter administration [96].

A PAMAM dendrimer hydrogel has been developed byHolden and coworkers that is made from “ultraviolet-curedPAMAMdendrimer” linkedwith PEG-acrylate chains for thedelivery of two antiglaucoma drugs which were brimonidine(0.1% w/v) and timolol maleate (0.5% w/v). Dendrimerichydrogel was obtained by crosslinking of the reactive acrylategroups, triggered by UV light. It was reported that thedendrimeric hydrogel was mucoadhesive and nontoxic to

epithelial cells of human cornea. Higher uptake from “humancorneal epithelial cells” and significantly enhanced “bovinecorneal transport” were reported for both drugs, comparedto the eye drops. The higher uptake in the dendrimerichydrogel formulations explained the temporary decomposi-tion of the corneal epithelial tight junctions [97]. The samegroup also developed a novel “hybrid PAMAM dendrimerhydrogel/poly(lactic-co-glycolic acid) (PLGA) nanoparticleplatform (HDNP)”, again for codelivery of brimonidine andtimolol maleate. The formulations were also evaluated interms of their in vitro potential toxicity and it was found thatthe formulation was noncytotoxic to human corneal epithe-lial cells. Following topical administration of the HDNP inadult “normotensive Dutch-beltedmale rabbits,” formulationwas found to be effective and maintained significantly higherconcentrations of both drugs up to 7 days in aqueous humorand cornea compared to saline. Furthermore, it was reportedthat dendrimeric hydrogel and PLGA nanoparticles were notinducing any ocular inflammation or discomfort. This studydemonstrated that this new formulation is able to enhancedrug bioavailability, and following topical administration,it is capable of sustaining drug activity [98]. Wathier andcoworkers also developed an in situ gel formulation usingLsyxCysy dendritic polymers to be used in cataract incisionsinstead of nylon sutures. It was reported that the hydrogelsealant procedure was simple and required less surgical timethan conventional suturing and no additional tissue traumawas inflicted [99].

7. Conclusion

Effective treatment of ocular diseases is still a challenge inpharmaceutical research, because of the unique physiologyof eye and presence of the ocular barriers especially inposterior segments of the eye. Research in ophthalmic drugdelivery during the last two decades has undergone majoradvancements from the use of conventional formulationssuch as solutions, suspensions, and ointments to viscosity-enhancing in situ gel systems, different inserts, colloidalsystems, and so forth.

Given their structural features,most of the ocular diseaseswould benefit from long-lasting drug delivery of dendrimersand dendrimer-based drug delivery systems. It was alreadyreported that dendrimers present practical solutions to drugdelivery issues such as solubility, biodistribution, and target-ing. Since it is easy to control the features of dendrimers suchas their size, shape, generation, branching length, molecularsize, and surface functionality, these compounds are idealcarriers in pharmaceutical applications. Recent researcheshave shown that dendrimers are able to

(i) enhance the corneal residence time of drugs admin-istered topically,

(ii) target retinal neuroinflammation and provide tar-geted, sustained neuroprotection in retinal degener-ation,

(iii) deliver drugs to the retina upon systemic administra-tion,

Page 10: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

10 The Scientific World Journal

(iv) be effective as corneal glues to potentially replacesutures following corneal surgeries.

Even though dendrimers are not approved yet for clinicaluse in the eye, their promising preclinical results can providesignificant opportunities.

Conflict of Interests

The authors would like to declare that they have no conflictof interests and have received no payment in the preparationof this paper.

References

[1] R. C. Nagarwal, S. Kant, P. N. Singh, P. Maiti, and J. K. Pandit,“Polymeric nanoparticulate system: a potential approach forocular drug delivery,” Journal of Controlled Release, vol. 136, no.1, pp. 2–13, 2009.

[2] R. Gaudana, J. Jwala, S. H. S. Boddu, and A. K. Mitra, “Recentperspectives in ocular drug delivery,” Pharmaceutical Research,vol. 26, no. 5, pp. 1197–1216, 2009.

[3] J. C. Lang, “Ocular drug delivery conventional ocular formula-tions,”Advanced Drug Delivery Reviews, vol. 16, no. 1, pp. 39–43,1995.

[4] V. P. Ranta, E. Mannermaa, K. Lummepuro et al., “Barrier ana-lysis of periocular drug delivery to the posterior segment,” Jour-nal of Controlled Release, vol. 148, no. 1, pp. 42–48, 2010.

[5] E. Eljarrat-Binstock, J. Pe’er, and A. J. Domb, “New techniquesfor drug delivery to the posterior eye segment,” PharmaceuticalResearch, vol. 27, no. 4, pp. 530–543, 2010.

[6] E. M. Del Amo and A. Urtti, “Current and future ophthalmicdrug delivery systems: a shift to the posterior segment,” DrugDiscovery Today, vol. 13, no. 3-4, pp. 135–143, 2008.

[7] V. P. Ranta and A. Urtti, “Transscleral drug delivery to the pos-terior eye: prospects of pharmacokinetic modeling,” AdvancedDrug Delivery Reviews, vol. 58, no. 11, pp. 1164–1181, 2006.

[8] A. Quintana, E. Raczka, L. Piehler et al., “Design and functionof a dendrimer-based therapeutic nanodevice targeted to tumorcells through the folate receptor,” Pharmaceutical Research, vol.19, no. 9, pp. 1310–1316, 2002.

[9] Y. Diebold and M. Calonge, “Applications of nanoparticles inophthalmology,” Progress in Retinal and Eye Research, vol. 29,no. 6, pp. 596–609, 2010.

[10] T. Yasukawa, Y. Ogura, Y. Tabata, H. Kimura, P. Wiedemann,andY.Honda, “Drugdelivery systems for vitreoretinal diseases,”Progress in Retinal and Eye Research, vol. 23, no. 3, pp. 253–281,2004.

[11] M.Campbell, A. T.H.Nguyen,A. S. Kiang et al., “Reversible andsize-selective opening of the inner blood-retina barrier: anovel therapeutic strategy,” in Retinal Degenerative Diseases:Laboratory andTherapeutic Investigations, vol. 664, pp. 301–308,Springer, New York, NY, USA, 2010.

[12] P. Pahuja, S. Arora, and P. Pawar, “Ocular drug delivery system:a reference to natural polymers,” Expert Opinion on DrugDelivery, vol. 9, no. 7, pp. 837–861, 2012.

[13] S. Wadhwa, R. Paliwal, S. R. Paliwal, and S. P. Vyas, “Nanocar-riers in ocular drug delivery: an update review,” CurrentPharmaceutical Design, vol. 15, no. 23, pp. 2724–2750, 2009.

[14] S. Raghava, M. Hammond, and U. B. Kompella, “Periocularroutes for retinal drug delivery,” Expert Opinion on DrugDelivery, vol. 1, no. 1, pp. 99–114, 2004.

[15] P. B. Patel, D. H. Shastri, P. K. Shelat, and A. K. Shukla, “Oph-thalmic drug delivery system: challenges and approaches,”Systematic Reviews in Pharmacy, vol. 1, no. 2, pp. 113–120, 2010.

[16] H. D. Perry, R. Solomon, E. D. Donnenfeld et al., “Evaluationof topical cyclosporine for the treatment of dry eye disease,”Archives of Ophthalmology, vol. 126, no. 8, pp. 1046–1050, 2008.

[17] B. Yavuz, S. Bozdag Pehlıvan, andN. Unlu, “An overview on dryeye treartment: approaches for cyclosporin a delivery,” TheScientific World Journal, vol. 2012, pp. 1–12, 2012.

[18] G. Venkata Ratnam, S. Madhavi, and P. Rajesh, “Ocular drugdelivery: an update review,” International Journal of Pharma andBio Sciences, vol. 1, no. 4, pp. 437–446, 2011.

[19] M. Hamidi, A. Azadi, and P. Rafiei, “Hydrogel nanoparticles indrug delivery,” Advanced Drug Delivery Reviews, vol. 60, no. 15,pp. 1638–1649, 2008.

[20] J. Jiang, J. S. Moore, H. F. Edelhauser, andM. R. Prausnitz, “Intr-ascleral drug delivery to the eye using hollow microneedles,”Pharmaceutical Research, vol. 26, no. 2, pp. 395–403, 2009.

[21] A. Urtti, “Challenges and obstacles of ocular pharmacokineticsand drug delivery,”Advanced Drug Delivery Reviews, vol. 58, no.11, pp. 1131–1135, 2006.

[22] S. K. Sahoo, F. Diinawaz, and S. Krishnakumar, “Nanotechnol-ogy in ocular drug delivery,” Drug Discovery Today, vol. 13, no.3-4, pp. 144–151, 2008.

[23] E. R. Gillies and J. M. Frechet, “Dendrimers and dendriticpolymers in drug delivery,” Drug Discovery Today, vol. 10, no.1, pp. 35–43, 2005.

[24] Y. Y. Cheng, Z. H. Xu, M. L. Ma, and T. W. Xu, “Dendrimers asdrug carriers: applications in different routes of drug admin-istration,” Journal of Pharmaceutical Sciences, vol. 97, no. 1, pp.123–143, 2008.

[25] R. Esfand and D. A. Tomalia, “Poly(amidoamine) (pamam)dendrimers: from biomimicry to drug delivery and biomedicalapplications,” Drug Discovery Today, vol. 6, no. 8, pp. 427–436,2001.

[26] S. Hecht and J. M. J. Frechet, “Dendritic encapsulation of func-tion: applying nature’s site isolation principle from biomimeticstomaterials science,”Angewandte Chemie International Edition,vol. 40, no. 1, pp. 74–91, 2001.

[27] V. Trivedi, U. Patel, B. Bhimani, D. Daslania, G. Patel, and B.Vyas, “Dendrimer: a polymer of 21st century,” IJPRBS Journal,vol. 1, no. 2, pp. 1–21, 2012.

[28] P. Hodge, “Organic-chemistry: polymer science branches out,”Nature, vol. 362, no. 6415, pp. 18–19, 1993.

[29] B. Klajnert and M. Bryszewska, “Dendrimers: properties andapplications,” Acta Biochimica Polonica, vol. 48, no. 1, pp. 199–208, 2001.

[30] B. K. Nanjwade, H. M. Bechra, G. K. Derkar, F. V. Manvi, and V.K. Nanjwade, “Dendrimers: emerging polymers for drug-delivery systems,” European Journal of Pharmaceutical Sciences,vol. 38, no. 3, pp. 185–196, 2009.

[31] D. A. Tomalia, “Birth of a new macromolecular architecture:dendrimers as quantized building blocks for nanoscale syn-thetic polymer chemistry,” Progress in Polymer Science, vol. 30,no. 3-4, pp. 294–324, 2005.

[32] J. P. Bharti, S. K. Prajapati, M. K. Jaiswal, and R. D. Yadav,“Dendrimer multifunctional nano-device: a review,” Interna-tional Journal of Pharmaceutical Sciences and Research, vol. 2,no. 8, pp. 1947–1960, 2011.

Page 11: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 11

[33] T. Garg, O. Singh, S. Arora, and R. Murthy, “Dendrimer: anovel scaffold for drug delivery,” International Journal of Phar-maceutical Sciences Review and Research, vol. 7, no. 2, pp. 211–220, 2011.

[34] L. M. Kaminskas, B. J. Boyd, and C. J. H. Porter, “Dendrimerpharmacokinetics: the effect of size, structure and surfacecharacteristics on adme properties,” Nanomedicine, vol. 6, no.6, pp. 1063–1084, 2011.

[35] R. Jevprasesphant, J. Penny,D.Attwood,N. B.McKeown, andA.D’Emanuele, “Engineering of dendrimer surfaces to enhancetransepithelial transport and reduce cytotoxicity,” Pharmaceu-tical Research, vol. 20, no. 10, pp. 1543–1550, 2003.

[36] H. T. Chen, M. F. Neerman, A. R. Parrish, and E. E. Simanek,“Cytotoxicity, hemolysis, and acute in vivo toxicity of den-drimers based on melamine, candidate vehicles for drug deliv-ery,” Journal of the American Chemical Society, vol. 126, no. 32,pp. 10044–10048, 2004.

[37] K. Jain, P. Kesharwani, U. Gupta, and N. K. Jain, “Dendrimertoxicity: let’s meet the challenge,” International Journal ofPharmaceutics, vol. 394, no. 1-2, pp. 122–142, 2010.

[38] S. Shaunak, S. Thomas, E. Gianasi et al., “Polyvalent dendrimerglucosamine conjugates prevent scar tissue formation,” NatureBiotechnology, vol. 22, no. 8, pp. 977–984, 2004.

[39] S. J. Kang, C. Durairaj, U. B. Kompella, J. M. O’Brien, and H. E.Grossniklaus, “Subconjunctival nanoparticle carboplatin in thetreatment of murine retinoblastoma,” Archives of Ophthalmol-ogy, vol. 127, no. 8, pp. 1043–1047, 2009.

[40] J. D. Eichman, A. U. Bielinska, J. F. Kukowska-Latallo, and J. R.Baker Jr., “The use of pamam dendrimers in the efficienttransfer of genetic material into cells,” Pharmaceutical Scienceand Technology Today, vol. 3, no. 7, pp. 232–245, 2000.

[41] N. P. Silva, F. P. Menacho, and M. Chorilli, “Dendrimers aspotential platform in nanotechnology-based drug delivery sys-tems,” IOSR Journal of Pharmacy, vol. 2, no. 5, pp. 23–30, 2012.

[42] D. A. Tomalia, “Starburst dendrimers: molecular-level controlof size, shape, surface chemistry, topology, and flexibility fromatoms to macroscopic matter,” Angewandte Chemie Interna-tional Edition, vol. 29, no. 2, pp. 138–175, 1990.

[43] R. Duncan and L. Izzo, “Dendrimer biocompatibility andtoxicity,” Advanced Drug Delivery Reviews, vol. 57, no. 15, pp.2215–2237, 2005.

[44] A.Malik, S. Chaudhary, G. Garg, and A. Tomar, “Dendrimers: atool for drug delivery,” Advances in Biological Research, vol. 6,no. 4, pp. 165–169, 2012.

[45] N.Malik, R.Wiwattanapatapee, R. Klopsch et al., “Dendrimers:relationship between structure and biocompatibility in vitroand preliminary studies on the biodistribution of i-125-labelledpolyamidoamine dendrimers in vivo,” Journal of ControlledRelease, vol. 65, no. 1-2, pp. 133–148, 2000.

[46] M. Liu, K. Kono, and J. M. Frechet, “Water-soluble dendriticunimolecular micelles: their potential as drug delivery agents,”Journal of Controlled Release, vol. 65, no. 1-2, pp. 121–131, 2000.

[47] J. M. Oliveira, A. J. Salgado, N. Sousa, J. F. Mano, and R. L. Reis,“Dendrimers and derivatives as a potential therapeutic tool inregenerative medicine strategies: a review,” Progress in PolymerScience, vol. 35, no. 9, pp. 1163–1194, 2010.

[48] S. H. Medina and M. E. El-Sayed, “Dendrimers as carriers fordelivery of chemotherapeutic agents,” Chemical Reviews, vol.109, no. 7, pp. 3141–3157, 2009.

[49] V. Gajbhiye, P. Vijayaraj Kumar, R. K. Tekade, and N. K. Jain,“Pegylated ppi dendritic architectures for sustained delivery of

h2 receptor antagonist,” European Journal of Medicinal Chem-istry, vol. 44, no. 3, pp. 1155–1166, 2009.

[50] N. A. Stasko, C. B. Johnson, M. H. Schoenfisch, T. A. Johnson,and E. L. Holmuhamedov, “Cytotoxicity of polypropyleniminedendrimer conjugates on cultured endothelial cells,” Biomacro-molecules, vol. 8, no. 12, pp. 3853–3859, 2007.

[51] P. Kumar, K. P. Meena, P. Kumar, C. Choudhary, D. S. Thakur,and P. Bajpayee, “Dendrimer: a novel polymer for drug deliv-ery,” JITPS Journal, vol. 1, no. 6, pp. 252–269, 2010.

[52] A. R. Menjoge, R. M. Kannan, and D. A. Tomalia, “Dendrimer-based drug and imaging conjugates: design considerations fornanomedical applications,”Drug Discovery Today, vol. 15, no. 5-6, pp. 171–185, 2010.

[53] T. D.McCarthy, P. Karellas, S. A. Henderson et al., “Dendrimersas drugs: discovery and preclinical and clinical developmentof dendrimer-based microbicides for hiv and sti prevention,”Molecular Pharmaceutics, vol. 2, no. 4, pp. 312–318, 2005.

[54] W. Wijagkanalan, S. Kawakami, and M. Hashida, “Designingdendrimers for drug delivery and imaging: pharmacokineticconsiderations,” Pharmaceutical Research, vol. 28, no. 7, pp.1500–1519, 2011.

[55] A. D’Emanuele and D. Attwood, “Dendrimer-drug interac-tions,”Advanced Drug Delivery Reviews, vol. 57, no. 15, pp. 2147–2162, 2005.

[56] N. K. Jain and U. Gupta, “Application of dendrimer-drug com-plexation in the enhancement of drug solubility and bioavail-ability,” Expert Opinion onDrugMetabolism and Toxicology, vol.4, no. 8, pp. 1035–1052, 2008.

[57] D. K. Smith, “Dendritic gels: many armsmake light work,”Adv-anced Materials, vol. 18, no. 20, pp. 2773–2778, 2006.

[58] D. L. Richter-Egger, A. Tesfai, and S. A. Tucker, “Spectrosco-pic investigations of poly(propyleneimine)dendrimers usingthe solvatochromic probe phenol blue and comparisons topoly(amidoamine) dendrimers,” Analytical Chemistry, vol. 73,no. 23, pp. 5743–5751, 2001.

[59] J. F. G. A. Jansen, E. M. M. Debrabandervandenberg, and E. W.Meijer, “Encapsulation of guest molecules into a dendritic box,”Science, vol. 266, no. 5188, pp. 1226–1229, 1994.

[60] J. F. G. A. Jansen JFGA, E. M. M. Debrabandervandenberg, andE. W. Meijer, “The dendritic box and bengal rose,” PolymericMaterials, vol. 73, pp. 123–124, 1995.

[61] J. F. G. A. Jansen, E.W.Meijer, and E. M.M. Debrabandervand-enberg, “The dendritic box: shape-selective liberation of encap-sulated guests,” Journal of the American Chemical Society, vol.117, no. 15, pp. 4417–4418, 1995.

[62] C. Kojima, K. Kono, K. Maruyama, and T. Takagishi, “Synthesisof polyamidoamine dendrimers having poly(ethylene glycol)grafts and their ability to encapsulate anticancer drugs,” Biocon-jugate Chemistry, vol. 11, no. 6, pp. 910–917, 2000.

[63] K. Kono,M. Liu, and J.M. J. Frechet, “Design of dendritic macr-omolecules containing folate or methotrexate residues,” Biocon-jugate Chemistry, vol. 10, no. 6, pp. 1115–1121, 1999.

[64] S. Stevelmans, J. C. M. vanHest, J. F. G. A. Jansen, D. A. F. J.vanBoxtel, E. M.M. D. Vanden Berg, and E.W.Meijer, “Synthe-sis, characterization and guest-host properties of inverted uni-molecular dendriticmicelles,” Journal of the American ChemicalSociety, vol. 118, no. 31, pp. 7398–7399, 1996.

[65] G. F. Pan, Y. Lemmouchi, E.O.Akala, andO. Bakare, “Studies onpegylated and drug-loaded pamam dendrimers,” Journal ofBioactive and Compatible Polymers, vol. 20, no. 1, pp. 113–128,2005.

Page 12: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

12 The Scientific World Journal

[66] R. B. Greenwald, Y. H. Choe, J. McGuire, and C. D. Cono-ver, “Effective drug delivery by pegylated drug conjugates,”Advanced Drug Delivery Reviews, vol. 55, no. 2, pp. 217–250,2003.

[67] M. S.Wendland and S. C. Zimmerman, “Synthesis of cored den-drimers,” Journal of the American Chemical Society, vol. 121, no.6, pp. 1389–1390, 1999.

[68] L. G. Schultz, Y. Zhao, and S. C. Zimmerman, “Synthesis ofcored dendrimers with internal cross-links,” Angewandte Che-mie International Edition, vol. 40, no. 10, pp. 1962–1966, 2001.

[69] T. Kim, H. J. Seo, J. S. Choi et al., “Pamam-peg-pamam: noveltriblock copolymer as a biocompatible and efficient gene deliv-ery carrier,” Biomacromolecules, vol. 5, no. 6, pp. 2487–2492,2004.

[70] U. Boas and P. M. Heegaard, “Dendrimers in drug research,”Chemical Society Reviews, vol. 33, no. 1, pp. 43–63, 2004.

[71] S. El Kazzouli, S. Mignani, M. Bousmina, and J. P. Majoral,“Dendrimer therapeutics: covalent and ionic attachments,”NewJournal of Chemistry, vol. 36, no. 2, pp. 227–240, 2012.

[72] A. K. Patri, J. F. Kukowska-Latallo, and J. R. Baker, “Targeteddrug delivery with dendrimers: comparison of the release kine-tics of covalently conjugated drug and non-covalent druginclusion complex,”AdvancedDrugDelivery Reviews, vol. 57, no.15, pp. 2203–2214, 2005.

[73] L. M. Kaminskas, V. M. McLeod, C. J. Porter, and B. J. Boyd,“Association of chemotherapeutic drugs with dendrimernanocarriers: an assessment of the merits of covalentconjugation compared to noncovalent encapsulation,” Molecu-lar Pharmaceutics, vol. 9, no. 3, pp. 355–373, 2012.

[74] T. R. Hoare and D. S. Kohane, “Hydrogels in drug delivery: pro-gress and challenges,” Polymer, vol. 49, no. 8, pp. 1993–2007,2008.

[75] H. B. Nirmal, S. R. Bakliwal, and S. P. Pawar, “In-situ gel: newtrends in controlled and sustained drug delivery system,” Inter-national Journal of PharmTech Research, vol. 2, no. 2, pp. 1398–1408, 2010.

[76] R. S. Navath, A. R. Menjoge, H. Dai, R. Romero, S. Kannan, andR.M. Kannan, “Injectable pamam dendrimer-peg hydrogels forthe treatment of genital infections: formulation and in vitro andin vivo evaluation,” Molecular Pharmaceutics, vol. 8, no. 4, pp.1209–1223, 2011.

[77] S. H. M. Sontjens, D. L. Nettles, M. A. Carnahan, L. A. Setton,and M. W. Grinstaff, “Biodendrimer-based hydrogel scaffoldsfor cartilage tissue repair,” Biomacromolecules, vol. 7, no. 1, pp.310–316, 2006.

[78] W. D. Jang and T. Aida, “Dendritic physical gels: structuralparameters for gelation with peptide-core dendrimers,”Macro-molecules, vol. 36, no. 22, pp. 8461–8469, 2003.

[79] A. Marco Zarbin, F. James Leary, C. Montemagno, R. Ritch, andM. S. Humayun, “Nanomedicine in ophthalmology,” in Retina,S. J. Ryan, Ed., pp. 689–711, 2013.

[80] O. P. Perumal, R. Inapagolla, S. Kannan, andR.M.Kannan, “Theeffect of surface functionality on cellular trafficking of den-drimers,” Biomaterials, vol. 29, no. 24-25, pp. 3469–3476, 2008.

[81] S. P. Kambhampati and R. M. Kannan, “Dendrimer nanoparti-cles for ocular drug delivery,” Journal of Ocular PharmacologyandTherapeutics, vol. 29, no. 2, pp. 151–165, 2013.

[82] T. F. Vandamme and L. Brobeck, “Poly(amidoamine) den-drimers as ophthalmic vehicles for ocular delivery of pilo-carpine nitrate and tropicamide,” Journal of Controlled Release,vol. 102, no. 1, pp. 23–38, 2005.

[83] G. G. Spataro, F. Malecaze, C.-O. Turrin et al., “Designingdendrimers for ocular drug delivery,” European Journal ofMedicinal Chemistry, vol. 45, no. 1, pp. 326–334, 2010.

[84] C. Durairaj, R. S. Kadam, J. W. Chandler, S. L. Hutcherson, andU. B. Kompella, “Nanosized dendritic polyguanidilyated trans-locators for enhanced solubility, permeability and delivery ofgatifloxacin,” Investigative Ophthalmology and Visual Science,vol. 51, no. 11, pp. 5804–5816, 2010.

[85] R. J. Marano, I. Toth, N. Wimmer, M. Brankov, and P. E. Rako-czy, “Dendrimer delivery of an anti-vegf oligonucleotide intothe eye: a long-term study into inhibition of laser-induced cnv,distribution, uptake and toxicity,” Gene Therapy, vol. 12, no. 21,pp. 1544–1550, 2005.

[86] X. Duan and H. Sheardown, “Dendrimer crosslinked collagenas a corneal tissue engineering scaffold: mechanical propertiesand corneal epithelial cell interactions,”Biomaterials, vol. 27, no.26, pp. 4608–4617, 2006.

[87] X. D. Duan, C. McLaughlin, M. Griffith, and H. Sheardown,“Biofunctionalization of collagen for improved biologicalresponse: scaffolds for corneal tissue engineering,”Biomaterials,vol. 28, no. 1, pp. 78–88, 2007.

[88] M.W. Grinstaff, “Designing hydrogel adhesives for corneal wo-und repair,” Biomaterials, vol. 28, no. 35, pp. 5205–5214, 2007.

[89] K. Sugisaki, T. Usui, N. Nishiyama et al., “Photodynamic ther-apy for corneal neovascularization using polymeric micellesencapsulating dendrimer porphyrins,” Investigative Ophthal-mology and Visual Science, vol. 49, no. 3, pp. 894–899, 2008.

[90] A. Makky, J. P. Michel, P. Maillard, and V. Rosilio, “Biomimeticliposomes and planar supported bilayers for the assessment ofglycodendrimeric porphyrins interaction with an immobilizedlectin,” Biochimica et Biophysica Acta, vol. 1808, no. 3, pp. 656–666, 2011.

[91] N. Nishiyama, H. R. Stapert, G. D. Zhang et al., “Light-har-vesting ionic dendrimer porphyrins as new photosensitizers forphotodynamic therapy,” Bioconjugate Chemistry, vol. 14, no. 1,pp. 58–66, 2003.

[92] N. Nishiyama, Y. Morimoto, W. D. Jang, and K. Kataoka, “Des-ign and development of dendrimer photosensitizer-incorpo-rated polymeric micelles for enhanced photodynamic therapy,”Advanced Drug Delivery Reviews, vol. 61, no. 4, pp. 327–338,2009.

[93] N. Nishiyama, A. Iriyama,W.D. Jang et al., “Light-induced genetransfer from packaged DNA enveloped in a dendrimericphotosensitizer,” Nature Materials, vol. 4, no. 12, pp. 934–941,2005.

[94] I. Bravo-Osuna, R. Herrero-Vanrell, I. T. MolinaMartınez et al.,“In vitro and in vivo tolerance studies of carbosilane dendrimersfor ophthalmic administration,” Investigative Ophthalmologyand Visual Science, vol. 51, E-Abstract 437, 2010.

[95] W. J. Yao, K. X. Sun, H. J. Mu et al., “Preparation and characteri-zation of puerarindendrimer complexes as an ocular drug deli-very system,” Drug Development and Industrial Pharmacy, vol.36, no. 9, pp. 1027–1035, 2010.

[96] R. Iezzi, B. R. Guru, I. V. Glybina, M. K. Mishra, A. Kennedy,and R. M. Kannan, “Dendrimer-based targeted intravitreal the-rapy for sustained attenuation of neuroinflammation in retinaldegeneration,” Biomaterials, vol. 33, no. 3, pp. 979–988, 2012.

[97] C. A. Holden, P. Tyagi, A. Thakur et al., “Polyamidoamine den-drimer hydrogel for enhanced delivery of antiglaucoma drugs,”Nanomedicine, vol. 8, no. 5, pp. 776–783, 2012.

[98] H. Yang, P. Tyagi, R. S. Kadam, C. A. Holden, and U. B. Kom-pella, “Hybrid dendrimer hydrogel/plga nanoparticle platform

Page 13: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

The Scientific World Journal 13

sustains drug delivery for one week and antiglaucoma effectsfor four days following one-time topical administration,” ACSNano, vol. 6, no. 9, pp. 7595–7606, 2012.

[99] M. Wathier, P. J. Jung, M. A. Carnahan, T. Kim, and M. W. Gri-nstaff, “Dendritic macromers as in situ polymerizing biomate-rials for securing cataract incisions,” Journal of the AmericanChemical Society, vol. 126, no. 40, pp. 12744–12745, 2004.

Page 14: Review Article Dendrimeric Systems and Their Applications ...downloads.hindawi.com/journals/tswj/2013/732340.pdf · Review Article Dendrimeric Systems and Their Applications in Ocular

Submit your manuscripts athttp://www.hindawi.com

PainResearch and TreatmentHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com

Volume 2014

ToxinsJournal of

VaccinesJournal of

Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

AntibioticsInternational Journal of

ToxicologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

StrokeResearch and TreatmentHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Drug DeliveryJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Advances in Pharmacological Sciences

Tropical MedicineJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Medicinal ChemistryInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

AddictionJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

Emergency Medicine InternationalHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Autoimmune Diseases

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Anesthesiology Research and Practice

ScientificaHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Pharmaceutics

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of